Sei sulla pagina 1di 7

Neurocrit Care

https://doi.org/10.1007/s12028-019-00829-x

VIEWPOINT

High‑Dose Intravenous Ascorbic Acid: Ready


for Prime Time in Traumatic Brain Injury?
Stefan W. Leichtle1*  , Anand K. Sarma2, Micheal Strein3, Vishal Yajnik4, Dennis Rivet5, Adam Sima6
and Gretchen M. Brophy3,5

© 2019 Springer Science+Business Media, LLC, part of Springer Nature and Neurocritical Care Society

Abstract 
Traumatic brain injury (TBI) is one of the leading public health problems in the USA and worldwide. It is the num-
ber one cause of death and disability in children and adults between ages 1–44. Despite efforts to prevent TBIs, the
incidence continues to rise. Secondary brain injury occurs in the first hours and days after the initial impact and is
the most effective target for intervention. Inflammatory processes and oxidative stress play an important role in the
pathomechanism of TBI and are exacerbated by impaired endogenous defense mechanisms, including depletion of
antioxidants. As a reducing agent, free radical scavenger, and co-factor in numerous biosynthetic reactions, ascorbic
acid (AA, vitamin C) is an essential nutrient that rapidly becomes depleted in states of critical illness. The administra-
tion of high-dose intravenous (IV) AA has demonstrated benefits in numerous preclinical models in the areas of
trauma, critical care, wound healing, and hematology. A safe and inexpensive treatment, high-dose IV AA adminis-
tration gained recent attention in studies demonstrating an associated mortality reduction in septic shock patients.
High-quality data on the effects of high-dose IV AA on TBI are lacking. Historic data in a small number of patients
demonstrate acute and profound AA deficiency in patients with central nervous system pathology, particularly TBI,
and a strong correlation between low AA concentrations and poor outcomes. While replenishing deficient AA stores
in TBI patients should improve the brain’s ability to tolerate oxidative stress, high-dose IV AA may prove an effective
strategy to prevent or mitigate secondary brain injury due to its ability to impede lipid peroxidation, scavenge reac-
tive oxygen species, suppress inflammatory mediators, stabilize the endothelium, and reduce brain edema. The exist-
ing preclinical data and limited clinical data suggest that high-dose IV AA may be effective in lowering oxidative stress
and decreasing cerebral edema. Whether this translates into improved clinical outcomes will depend on identifying
the ideal target patient population and possible treatment combinations, factors that need to be evaluated in future
clinical studies. With its excellent safety profile and low cost, high-dose IV AA is ready to be evaluated in the early
treatment of TBI patients to mitigate secondary brain injury and improve outcomes.
Keywords:  Traumatic brain injury, Secondary brain injury, Oxidative stress, Antioxidant, Free radical scavenger,
Vitamin C, Ascorbic acid

*Correspondence: stefan.leichtle@vcuhealth.org
1
Division of Acute Care Surgical Services, Department of Surgery, Virginia
Commonwealth University School of Medicine, Richmond, USA
Full list of author information is available at the end of the article
Traumatic Brain Injury concentrations to above 1000  μmol/L, the optimal con-
Traumatic brain injury (TBI) is one of the leading pub- centration for free radical scavenging effects [20, 22-24].
lic health problems in the USA and worldwide, result- In 24 critically ill patients with sepsis, Fowler et al. dem-
ing in 2.8 million Emergency Department (ED) visits onstrated that IV administration of 200 mg/kg per day (as
and 56,000 deaths per year [1]. Already the leading cause compared to 50 mg/kg/d) in four bolus doses effectively
of death and disability in children and adults between and consistently elevated steady-state plasma concentra-
ages 1–44, the incidence of TBI continues to rise [1]. tions to above 1000 μmol/L within 24 h of the first dose
In addition to the direct brain injury caused by trau- [23]. Therefore, a treatment regimen of 50 mg/kg IV AA
matic impact, secondary brain injury due to local tissue given every 6 h (for a total of 200 mg/kg per day) is most
ischemia, edema, metabolic changes, and damage from likely to result in consistently supra-normal, ROS-scav-
reactive oxygen species (ROS) results in further neuro- enging plasma AA concentrations in critically ill patients.
logic deterioration [2-10]. Current treatment strategies to In its oxidized form dehydroascorbic acid (DHA), AA is
mitigate secondary brain injury are limited to the optimi- actively and preferentially transported into the central
zation of a patient’s general clinical condition, including nervous system (CNS), crossing the blood–brain barrier
maintenance of adequate cerebral perfusion pressures, (BBB) by facilitative transport. This mechanism results
management of intracranial hypertension, and preven- in substantially higher AA concentrations in the CNS
tion of hypotension, hypoxia, and seizures. No proven, than in plasma [25, 26]. Therefore, plasma concentrations
effective treatment exists to provide neuroprotection in of AA may not be reflective of CNS concentrations, but
the acute phase or to promote neuroregeneration in the their correlation is unclear.
delayed therapeutic window after TBI [11, 12]. AA administration has few clinically relevant adverse
effects. As a highly water-soluble compound, AA is
Ascorbic Acid (Vitamin C) readily excreted through kidneys and urine, limiting the
Ascorbic acid (AA, vitamin C) is an essential nutrient potential for systemic toxicity. Therefore, direct renal
that functions as an important reducing agent, free radi- toxicity and formation of oxalate stones is the most fre-
cal scavenger, and co-factor in numerous biosynthetic quently noted concern with high-dose AA treatment [27].
reactions [13-15]. The recommended dietary allow- Healthy individuals did not experience relevant adverse
ance for AA for healthy individuals is 75 mg and 90 mg effects or laboratory abnormalities with daily doses of
per day for women and men, respectively, though some 7500 mg IV for 6 days [28], critically ill patients tolerated
guidelines recommend higher doses of up to 200 mg per daily administration of 4500 mg IV for 28 days [29], and
day [16]. AA is readily absorbed from the intestinal tract even higher doses were found to be safe in phase-1 can-
when consumed orally, and higher doses above 1000 mg cer and sepsis trials [23, 30-32]. Occasionally reported
per day are excreted renally [17, 18]. While oral adminis- adverse effects include gastrointestinal discomfort and
tration of AA is adequate to maintain homeostasis, intake headache, but overall, high-dose IV AA is regarded to
of substantially higher doses is required to achieve supra- have a good safety profile with a low risk of toxicity over a
normal, ROS-scavenging AA concentrations, particularly wide range of doses.
in states of acute injury and critical illness [18, 19].
While not universally defined, normal AA concentra- Ascorbic Acid Deficiency in Critical Illness, Trauma,
tions range from 21 μmol/L (0.37 mg/dL) to 100 μmol/L and Burns
(1.76  mg/dL) [17, 20, 21]. In a small cohort of healthy Critical illness is marked by depletion of AA stores [33-
volunteers, mean peak plasma concentrations of AA 35]. The administration of antioxidants, including AA,
reached 134.8  ±  20.6  μmol/L after oral administra- in states of critical illness has re-emerged as a promising
tion of 1250  mg AA. In contrast, IV administration of area for study due to significant success in small patient
this dose resulted in peak plasma concentrations of cohorts with septic shock [23, 36] and supported by basic
885 ± 201.2  μmol/L. Predicted maximum peak plasma science research in trauma, wound healing, and hema-
concentrations were no more than 220 μmol/L with oral tology [37-40]. Traumatic injuries, acute infection, and
administration of 3000  mg AA every 4  h, but reached critical illness result in a pro-inflammatory state marked
1760  μmol/L for IV administration of this dose [22]. by dysregulation of the immune system, high levels of
Based on these and other data, any effective AA treat- ROS, and capillary leak due to endothelial damage and
ment regimen for critically ill patients must be admin- increased permeability [41-43]. Antioxidants, most
istered IV rather than orally. In 20 critically ill patients importantly AA, have the potential to influence these
with multiple organ dysfunction, 2000  mg AA per day pathomechanisms.
given IV restored normal AA plasma concentrations, but In rodent models of bacterial- or endotoxin-induced
only a daily dose of 10,000 mg IV AA elevated plasma AA sepsis, AA administration attenuated the inflammatory
response and decreased microvascular permeability, in acute illness due to the depletion of free radical scav-
decreasing their risk for organ dysfunction and mitigating engers such as AA [7, 8, 34, 50, 51]. Figure 1 shows one
lung damage [44, 45]. Clinically, lung-protective effects of of the proposed mechanisms of neuroprotection of high-
AA were described by Nathens et al. [29] in a cohort of dose IV AA in TBI. In a small cohort of patients with
severely injured trauma patients in the ICU setting. IV TBI and hemorrhagic stroke, plasma concentrations of
administration of 1000 mg AA three times per day, enter- AA were found to be low and inversely correlated with
ally supplemented by 1000 IU alpha-tocopherol, resulted injury severity. In this 2001 study, 13 patients with TBI
in modest but significant reduction of pulmonary com- had significantly lower plasma concentrations of AA
plications. Berger and colleagues studied a mix of antiox- (29 ± 8  μmol/L) than healthy controls (52 ± 8  μmol/L),
idant supplements (2700  mg AA per day, and selenium, and AA plasma concentrations inversely correlated with
zinc, and vitamin B1) in a randomized trial involving a hemorrhage size [34]. In a 1984 study of AA concentra-
heterogenous surgical population of 200 patients, 30% tions in the cerebrospinal fluid (CSF) of 41 patients with
of which were trauma patients. This antioxidant cocktail neurologic disorders, five patients with TBI had lower
reduced C-reactive protein (CRP) concentrations, but AA concentrations than all other patients, including
had no measurable effect on organ dysfunction [46]. those with hydrocephalus and seizure disorders. AA con-
In a porcine model of severe polytrauma and hemor- centrations in the CSF of patients with head and cervical
rhage, Reynolds et al. demonstrated that AA supplemen- trauma were 77 ± 56  μmol/L compared to the reference
tation mitigated end-organ damage on histopathologic cohort with an average of 203 ± 49 μmol/L [7].
assessment, severity of lung injury, and decreased levels These few clinical studies on AA deficiency in patients
of the pro-inflammatory markers interleukin (IL)-1β, with TBI are in contrast to more robust literature in ani-
IL-8, TNFα, plasminogen activator inhibitor-1, and tissue mal models. In rodent models of ischemic stroke, DHA
factor [37]. While similar effects have been corroborated exhibited neuroprotective effects in both pre-treated
in numerous prior small animal studies, there are no con- animals and those that received DHA after the ischemic
clusive data from clinical trials in (poly)trauma patients. insult [52, 53]. In a rodent model of TBI, combined
In animal and clinical studies of severe burn injuries, AA administration of moderate doses of AA and vitamin E
administration decreases capillary leak and significantly resulted in improved cellular defense mechanism and
reduces resuscitation fluid requirements [47-49]. Doses confirmed depletion of important antioxidants after TBI
evaluated ranged from 15  mg/kg/h in animal studies to [54]. In a rodent model of spinal cord injury, high-dose
66  mg/kg/h in clinical trials. Similar to other fields of AA administration significantly reduced tissue necrosis
study, animal studies far outnumber clinical trials, and and improved functional performance in rats [55].
results in the clinical setting are less impressive than in In addition to its potent action as free radical scav-
animal models. enger, AA may stabilize the endothelium and pro-
mote integrity of the BBB. Following the mechanical
Ascorbic Acid in TBI impact of TBI, ischemia, reperfusion, and the imbal-
Oxidative stress is an important contributor in the patho- ance between diminished protective factors such as
genesis of secondary brain injury [2, 4-10]. Cellular free radical scavengers and NF-E2 related factor-2
defense mechanisms against this damage are impaired [56, 57] and upregulated damaging factors such as

Fig. 1  Proposed reactive oxygen-scavenging-mechanism of high-dose IV ascorbic acid in TBI. TBI, traumatic brain injury; O
­ 2−, reactive oxygen spe-
cies; DNA, deoxyribonucleic acid
metalloproteinase (MMP)-9 lead to disruption of trans- The most appropriate dose of AA in TBI is not known.
membrane tight junction proteins in the basal lamina However, higher doses have shown benefit in other criti-
of the BBB [55, 56, 58-61]. ROS cause cell damage cally ill patients without causing harm. Therefore, mod-
through lipid peroxidation of the fatty acids in cell and erate to high doses of IV AA (10,000–20,000  mg per
mitochondrial membranes [62, 63]. With its potent day) may be required to replenish deficient AA stores in
antioxidant properties, high-dose AA may mitigate this patients with TBI experiencing oxidative stress several
cascade [57, 61]. days after injury. Due to the limited data on prevalence
Unfortunately, clinical data on the therapeutic use of and extent of AA deficiency in TBI, it is unclear if and
antioxidants to date have been inconclusive or disap- to what degree this would translate into protection from
pointing [64]. Strategies that showed promise in animal secondary brain injury and improved clinical outcomes.
models and preclinical testing such as mitochondrial Contemporary data on AA concentrations in plasma and
protection, progesterone administration, anticonvul- CSF of patients with varying degrees of TBI may allow for
sants, and several antioxidant strategies like resveratrol, the identification of a high-risk patient population that
flavonoids, and omega-3 polyunsaturated fatty acids have would benefit most from AA repletion and is currently
not translated into successful clinical trials [65-68]. Com- being studied by our research group. Based on animal
pared to other states of critical illness, there is a paucity of data [70, 71], the aging brain may be particularly suscep-
data on the clinical use of AA in TBI. Table 1 provides an tible to oxidative damage due to an inability to take up
overview of studies on AA in patients with critical illness and mobilize AA. Clinically, lower CNS/plasma AA ratio
in the last two decades, fewer than 10% of which included in older adults compared to younger patients with TBI
patients with severe TBI. In the only randomized con- may indicate a high-risk patient group for further study.
trolled trial on AA in TBI, high-dose (10,000 mg  days 1 Administration of high-dose IV AA (≥ 200  mg/kg per
and 4; 4000 mg days 5 through 7) IV AA administration day) would harness the anti-inflammatory, ROS-scav-
in 23 patients resulted in decreased perilesional edema enging, and cell membrane-stabilizing properties of AA.
on CT imaging (p = 0.01), but no improvement in out- These mechanisms of action may be most beneficial in
comes [69]. Due to the dosing regimen, it is unlikely that patients with significant cerebral edema and elevated
ROS-scavenging plasma levels were reached in this trial. intracranial pressure when administered within hours

Table 1  Pertinent clinical trials of IV ascorbic acid in the last two decades
References Design Type of injury Patients ­ osinga Results
Ascorbic acid d

Marik et al. 2017 [36] Retrospective before- Severe sepsis/septic 94 adults 6 g ×  4 ­daysb Mortality reduction
after shock
Fowler et al. 2014 [23] Randomized double- Severe sepsis (mostly 24 adults 50 mg/kg/day Reduced SOFA scores
blind placebo- respiratory) or 200 mg/kg/ and inflammatory
controlled day × 4 days markers
Razmkon et al. 2011 Randomized double- TBI with GCS ≤ 8 100 adults (four 10 g in day 1, 10 g on High-dose AA decreased
[69] blind placebo- groups, 23 patients day 4, 4 g daily for perilesional edema on
controlled received high-dose three more ­daysb head CT
AA)
Barbosa et al. 2009 [49] Randomized double- Burns with TBSA 32 children Up to 2.7 g ­dailyb Decreased lipid per-
blind placebo- 10-50%, at least (1.5 × upper daily oxidation and faster
controlled partial thickness intake based on age) wound healing
for 5 days
Berger 2008 [18] Randomized double- Complex cardiac sur- 200 adults (21 with 2.7 g for two days, 1.6 g Decreased inflammatory
blind placebo- gery, major trauma, SAH) for three ­daysb response (CRP)
controlled or SAH
Nathens et al. 2002 [29] Randomized prospec- Trauma patients, 595 patients 1 g TID for up to Decreased risk for MSOF,
tive trial excluding isolated 28 days or length of shorter ICU stay
TBI and patients with ICU ­stayb
GCS ≤ 6
Tanaka et al. 2000 [47] Randomized prospec- Burns with TBSA ≥ 20% 37 adults 66 mg/kg/hr as Decreased IVF require-
tive trial continuous infusion ments and improved
for 24 h respiratory function
CRP C-reactive protein, CT computed tomography, GCS Glasgow Coma Scale, IVF intravenous fluid, MSOF multi-system organ failure, SAH subarachnoid hemorrhage,
SOFA sequential organ failure assessment, TBI traumatic brain injury, TBSA total body surface area
a
  Only AA dosing shown
b
  AA was part of a treatment regimen including other medications
of TBI. Many animal studies that examined the admin- sepsis, doses of 200  mg/kg per day, given in four doses
istration of antioxidant cocktails and vitamins, includ- every 6  h, rapidly led to plasma AA concentrations of
ing AA, did not employ such high doses. However, these ≥ 1000  μmol/L [23]. In de Grooth et  al.’s study compar-
higher doses are required in critically ill patients to reli- ing low- vs. high-dose and intermittent vs. continuous
ably achieve protective antioxidant concentrations and administration of IV AA in septic patients, bolus dosing
beneficial clinical outcomes, as recently demonstrated in provided rapid plasma peak concentrations and continu-
patients with septic shock [23, 36]. ous dosing was effective in achieving high steady-state
Prehospital administration of IV AA to patients with concentrations [20]. For TBI patients in an ICU environ-
(suspected) TBI could be even more effective in mitigat- ment, both regimens of administration would be feasible.
ing damage to the blood–brain barrier and mitochondria
of the brain parenchyma, which starts to occur within
minutes of the traumatic event [62, 63]. However, pre- Conclusion
hospital administration of AA would add substantial Inflammatory processes, ROS, and endothelial dysfunc-
complexity to a clinical trial; challenges would include tion are well-established contributors to secondary brain
the correct identification of TBI as the cause for altered injury. The promising results achieved with antioxidants
mental status in (poly-)trauma patients, the possible and free radical scavengers in animal studies are in stark
lack of knowledge of comorbidities that could increase contrast to disappointing clinical results. Based on its
the risk profile of high-dose AA administration, and the mechanisms of action, animal data, and clinical results
logistical challenge of storing solutions of high-dose AA in critical illness, high-dose AA has exceptional poten-
on ambulances. For the latter, promising data exist on tial to be an effective treatment strategy in the acute
the stability of solutions containing low- to moderate AA phase of TBI. It may be able to mitigate secondary brain
doses [72]. If results of a solid clinical (in-hospital) trial injury due to its antioxidant, anti-inflammatory, and cell
indicate therapeutic efficacy of AA in TBI, studies on the membrane-stabilizing properties, while the low adverse
prehospital administration of AA for TBI in select patient effect and cost profile of AA make it well suited for clini-
populations should certainly be considered. cal studies in patients with TBI. A key challenge will be
to identify the ideal TBI patient population that is most
Limitations and Challenges likely to benefit from this treatment strategy. Based on
Primary TBI and ROS-mediated damage to brain paren- the existing data from related fields, patients with moder-
chyma and the BBB occur at the time of injury and ate to severe TBI, significant brain edema with elevated
shortly thereafter [62, 63], making administration of neu- intracranial pressure, and the older adult population may
roprotective agents challenging. Even in mature trauma represent ideal patient groups for high-dose IV AA in the
systems, the first dose of AA may realistically not occur acute setting. Clinical trials in such high-risk populations,
until hours after injury rather than minutes after the ini- rather than a more heterogenous patient group with TBI,
tial traumatic event as in preclinical models. Therefore, may be the key to translating the promising animal data
the effect of high-dose AA will likely be of particular ben- into actual improved patient outcomes. In summary, the
efit in those with areas of substantial ischemic penum- existing ample animal and limited clinical data suggest
bra, cerebral edema, and elevated intracranial pressure. that for the right patient population and at the appropri-
Additional benefit would be expected from the general ate dose, high-dose IV AA can benefit patients with TBI.
effect of high-dose AA on the state of TBI-related critical
illness. Author details
In rodent models of TBI, the oxidized form of AA, 1
 Division of Acute Care Surgical Services, Department of Surgery, Virginia
Commonwealth University School of Medicine, Richmond, USA. 2 Department
DHA, is more effective in crossing the BBB [52] and con- of Neurology, Wake Forest School of Medicine, Winston‑Salem, USA. 3 Depart-
centration-dependent downregulation of AA transport- ment of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth
ers across the BBB might impede the accumulation of University School of Pharmacy, Richmond, USA. 4 Division of Critical Care,
Department of Anesthesiology, Virginia Commonwealth University School
supra-normal, ROS-scavenging concentrations of AA in of Medicine, Richmond, USA. 5 Department of Neurosurgery, Virginia Com-
the CNS [73]. DHA is not currently FDA-approved and monwealth University School of Medicine, Richmond, USA. 6 Department
is only used in experimental models. However, the AA of Biostatistics, Virginia Commonwealth University, Richmond, USA.
plasma concentrations that can be achieved with high- Author Contributions
dose IV administration will provide a steep plasma con- This manuscript complies with all instructions to authors. Authorship require-
centration gradient [74] ensuring uptake into the CNS. ments have been met by all authors, and the final manuscript was approved
by all co-authors.
Lastly, both continuous and intermittent administration
regimens of AA may need to be examined. In Fowler Conflict of interest
et al.’s phase I study on high-dose IV AA in patients with The authors declare that they have no conflict of interest.
Publisher’s Note 23. Fowler AA, Syed AA, Knowlson S, et al. Phase I safety trial of intravenous
Springer Nature remains neutral with regard to jurisdictional claims in pub- ascorbic acid in patients with severe sepsis. J Transl Med. 2014;12:32.
lished maps and institutional affiliations. 24. Jackson TS, Xu A, Vita JA, Keaney JF. Ascorbate prevents the interaction
of superoxide and nitric oxide only at very high physiological concentra-
tions. Circ Res. 1998;83(9):916–22.
25. May JM. Vitamin C transport and its role in the central nervous system.
Subcell Biochem. 2012;56:85–103.
26. Harrison FE, May JM. Vitamin C function in the brain: vital role of the
ascorbate transporter SVCT2. Free Radic Biol Med. 2009;46(6):719–30.
References 27. Buehner M, Pamplin J, Studer L, et al. Oxalate nephropathy after continu-
1. Taylor CA, Bell JM, Breiding MJ, Xu L. Traumatic brain injury-related emer- ous infusion of high-dose vitamin c as an adjunct to burn resuscitation. J
gency department visits, hospitalizations, and deaths—United States, Burn Care Res. 2016;37(4):e374–379.
2007 and 2013. MMWR Surveill Summ. 2017;66(9):1–16. 28. Muhlhofer A, Mrosek S, Schlegel B, et al. High-dose intravenous vitamin C
2. Dearden NM. Mechanisms and prevention of secondary brain damage is not associated with an increase of pro-oxidative biomarkers. Eur J Clin
during intensive care. Clin Neuropathol. 1998;17(4):221–8. Nutr. 2004;58(8):1151–8.
3. Jones PA, Andrews PJ, Midgley S, et al. Measuring the burden of second- 29. Nathens AB, Neff MJ, Jurkovich GJ, et al. Randomized, prospective trial of
ary insults in head-injured patients during intensive care. J Neurosurg antioxidant supplementation in critically ill surgical patients. Ann Surg.
Anesthesiol. 1994;6(1):4–14. 2002;236(6):814–22.
4. Lok J, Leung W, Murphy S, Butler W, Noviski N, Lo EH. Intracranial hemor- 30. Stephenson CM, Levin RD, Spector T, Lis CG. Phase I clinical trial to evalu-
rhage: mechanisms of secondary brain injury. Acta Neurochir Suppl. ate the safety, tolerability, and pharmacokinetics of high-dose intrave-
2011;111:63–9. nous ascorbic acid in patients with advanced cancer. Cancer Chemother
5. Ikeda Y, Long DM. The molecular basis of brain injury and brain edema: Pharmacol. 2013;72(1):139–46.
the role of oxygen free radicals. Neurosurgery. 1990;27(1):1–11. 31. Hoffer LJ, Levine M, Assouline S, et al. Phase I clinical trial of i.v. ascorbic
6. Hall E, Braughler J. Central nervous system trauma and stroke: II. Physi- acid in advanced malignancy. Ann Oncol. 2008;19(11):1969–74.
ological and pharmacological evidence for involvement of oxygen 32. Monti DA, Mitchell E, Bazzan AJ, et al. Phase I evaluation of intravenous
radicals and lipid peroxidation. Free Rad Biol Med. 1989;6(3):303–13. ascorbic acid in combination with gemcitabine and erlotinib in patients
7. Brau RH, García-Castiñeiras S, Rifkinson N. Cerebrospinal fluid ascorbic with metastatic pancreatic cancer. PLoS ONE. 2012;7(1):e29794.
acid levels in neurological disorders. Neurosurgery. 1984;14(2):142–6. 33. Carr AC, Rosengrave PC, Bayer S, Chambers S, Mehrtens J, Shaw GM.
8. Tyurin VA, Tyurina YY, Borisenko GG, et al. Oxidative stress following trau- Hypovitaminosis C and vitamin C deficiency in critically ill patients
matic brain injury in rats: quantitation of biomarkers and detection of free despite recommended enteral and parenteral intakes. Crit Care.
radical intermediates. J Neurochem. 2002;75(5):2178–89. 2017;21(1):300.
9. Kontos HA, Povlishock JT. Oxygen radicals in brain injury. Central Nervous 34. Polidori MC, Mecocci P, Frei B. Plasma vitamin C levels are decreased and
System Trauma. 1986;3(4):257–63. correlated with brain damage in patients with intracranial hemorrhage or
10. Povlishock JT, Kontos HA. The role of oxygen radicals in the pathobiology head trauma. Stroke. 2001;32(4):898–902.
of traumatic brain injury. Hum Cell. 1992;5(4):345–53. 35. Luo M, Fernandez-Estivariz C, Jones DP, et al. Depletion of plasma antioxi-
11. Hutchinson PJ, Kolias AG, Timofeev IS, et al. Trial of decompressive dants in surgical intensive care unit patients requiring parenteral feeding:
craniectomy for traumatic intracranial hypertension. N Engl J Med. effects of parenteral nutrition with or without alanyl-glutamine dipeptide
2016;375(12):1119–30. supplementation. Nutrition. 2008;24(1):37–44.
12. Andrews PJD, Sinclair HL, Rodriguez A, et al. Hypothermia for intrac- 36. Marik PE, Khangoora V, Rivera R, Hooper MH, Catravas J. Hydrocortisone,
ranial hypertension after traumatic brain injury. N Engl J Med. vitamin C, and thiamine for the treatment of severe sepsis and septic
2015;373(25):2403–12. shock: a retrospective before-after study. Chest. 2017;151(6):1229–388.
13. DePhillipo NN, Aman ZS, Kennedy MI, Begley JP, Moatshe G, LaPrade RF. 37. Reynolds PS, Fisher BJ, McCarter J, et al. Interventional vitamin C: A
Efficacy of vitamin C supplementation on collagen synthesis and oxida- strategy for attenuation of coagulopathy and inflammation in a swine
tive stress after musculoskeletal injuries: a systematic review. Orthop J multiple injuries model. J Trauma Acute Care Surg. 2018;85(1S Suppl
Sports Med. 2018;6(10):2325967118804544. 2):S57–S67.
14. Biesalski HK, McGregor GP. Antioxidant therapy in critical care–is 38. Sanford K, Fisher BJ, Fowler E, Fowler AA, Natarajan R. Attenuation of
the microcirculation the primary target? Crit Care Med. 2007;35(9 red blood cell storage lesions with vitamin C. Antioxidants (Basel).
Suppl):S577–583. 2017;6(3):E55.
15. Ellis GR, Anderson RA, Lang D, et al. Neutrophil superoxide anion–gen- 39. Mohammed BM, Fisher BJ, Kraskauskas D, et al. Vitamin C promotes
erating capacity, endothelial function and oxidative stress in chronic wound healing through novel pleiotropic mechanisms. Int Wound J.
heart failure: effects of short- and long-term vitamin C therapy. J Am Coll 2016;13(4):572–84.
Cardiol. 2000;36(5):1474–82. 40. Barichello T, Machado RA, Constantino L, et al. Antioxidant treatment
16. Vanek VW, Borum P, Buchman A, et al. A.S.P.E.N. position paper: recom- prevented late memory impairment in an animal model of sepsis. Crit
mendations for changes in commercially available parenteral multivita- Care Med. 2007;35(9):2186–90.
min and multi-trace element products. Nutr Clin Pract. 2012;27(4):440–91. 41. Haywood-Watson RJ, Holcomb JB, Gonzalez EA, et al. Modulation of
17. Levine M, Conry-Cantilena C, Wang Y, et al. Vitamin C pharmacokinetics in syndecan-1 shedding after hemorrhagic shock and resuscitation. PLoS
healthy volunteers: evidence for a recommended dietary allowance. Proc ONE. 2011;6(8):e23530.
Natl Acad Sci USA. 1996;93(8):3704–9. 42. Johansson PI, Henriksen HH, Stensballe J, et al. Traumatic endotheliopa-
18. Berger MM. Vitamin C requirements in parenteral nutrition. Gastroenterol- thy: a prospective observational study of 424 severely injured patients.
ogy. 2009;137(5 Suppl):S70–78. Ann Surg. 2017;265(3):597–603.
19. Long CL, Maull KI, Krishnan RS, et al. Ascorbic acid dynamics in the seri- 43. Wei S, Gonzalez Rodriguez E, Chang R, et al. Elevated syndecan-1 after
ously ill and injured. J Surg Res. 2003;109(2):144–8. trauma and risk of sepsis: a secondary analysis of patients from the
20. de Grooth H-J, Manubulu-Choo W-P, Zandvliet AS, et al. Vitamin C pragmatic, randomized optimal platelet and plasma ratios (PROPPR) Trial.
pharmacokinetics in critically Ill patients: a randomized trial of four IV J Am Coll Surg. 2018;227(6):587–95.
regimens. Chest. 2018;153(6):1368–77. 44. Fisher BJ, Seropian IM, Kraskauskas D, et al. Ascorbic acid attenu-
21. Levine M, Rumsey SC, Daruwala R, Park JB, Wang Y. Criteria and recom- ates lipopolysaccharide-induced acute lung injury. Crit Care Med.
mendations for vitamin C intake. JAMA. 1999;281(15):1415–23. 2011;39(6):1454–60.
22. Padayatty SJ, Sun H, Wang Y, et al. Vitamin C pharmacokinetics: implica- 45. Fisher BJ, Kraskauskas D, Martin EJ, et al. Mechanisms of attenuation
tions for oral and intravenous use. Ann Intern Med. 2004;140(7):533–7. of abdominal sepsis induced acute lung injury by ascorbic acid. Am J
Physiol Lung Cell Mol Physiol. 2012;303(1):L20–32.
46. Berger MM, Soguel L, Shenkin A, et al. Influence of early antioxidant sup- 60. Guo M, Cox B, Mahale S, et al. Pre-ischemic exercise reduces matrix metal-
plements on clinical evolution and organ function in critically ill cardiac loproteinase-9 expression and ameliorates blood-brain barrier dysfunc-
surgery, major trauma, and subarachnoid hemorrhage patients. Crit Care. tion in stroke. Neuroscience. 2008;151(2):340–51.
2008;12(4):R101. 61. Lin J-L, Huang Y-H, Shen Y-C, Huang H-C, Liu P-H. Ascorbic acid prevents
47. Tanaka H, Matsuda T, Miyagantani Y, Yukioka T, Matsuda H, Shimazaki blood-brain barrier disruption and sensory deficit caused by sustained
S. Reduction of resuscitation fluid volumes in severely burned patients compression of primary somatosensory cortex. J Cereb Blood Flow
using ascorbic acid administration: a randomized, prospective study. Arch Metab. 2010;30(6):1121–36.
Surg. 2000;135(3):326–31. 62. Smith SL, Andrus PK, Zhang JR, Hall ED. Direct measurement of hydroxyl
48. Dubick MA, Williams C, Elgjo GI, Kramer GC. High-dose vitamin C infusion radicals, lipid peroxidation, and blood-brain barrier disruption follow-
reduces fluid requirements in the resuscitation of burn-injured sheep. ing unilateral cortical impact head injury in the rat. J Neurotrauma.
Shock. 2005;24(2):139–44. 1994;11(4):393–404.
49. Barbosa E, Faintuch J, Machado Moreira EA, et al. Supplementation of 63. Vagnozzi R, Marmarou A, Tavazzi B, et al. Changes of cerebral energy
vitamin E, vitamin C, and zinc attenuates oxidative stress in burned chil- metabolism and lipid peroxidation in rats leading to mitochondrial
dren: a randomized, double-blind, placebo-controlled pilot study. J Burn dysfunction after diffuse brain injury. J Neurotrauma. 1999;16(10):903–13.
Care Res. 2009;30(5):859–66. 64. Putzu A, Daems A-M, Lopez-Delgado JC, Giordano VF, Landoni G. The
50. Kochanek PM, Dixon CE, Shellington DK, et al. Screening of biochemical effect of vitamin C on clinical outcome in critically ill patients: a system-
and molecular mechanisms of secondary injury and repair in the brain atic review with meta-analysis of randomized controlled trials. Crit Care
after experimental blast-induced traumatic brain injury in rats. J Neuro- Med. 2019;47(6):774–83.
trauma. 2013;30(11):920–37. 65. Maas AIR, Murray G, Henney H, et al. Efficacy and safety of dexanabinol in
51. Arun P, Rittase WB, Wilder DM, Wang Y, Gist ID, Long JB. Defective methio- severe traumatic brain injury: results of a phase III randomised, placebo-
nine metabolism in the brain after repeated blast exposures might controlled, clinical trial. Lancet Neurol. 2006;5(1):38–45.
contribute to increased oxidative stress. Neurochem Int. 2018;112:234–8. 66. Marshall LF, Maas AI, Marshall SB, et al. A multicenter trial on the
52. Huang J, Agus DB, Winfree CJ, et al. Dehydroascorbic acid, a blood-brain efficacy of using tirilazad mesylate in cases of head injury. J Neurosurg.
barrier transportable form of vitamin C, mediates potent cerebroprotec- 1998;89(4):519–25.
tion in experimental stroke. Proc Natl Acad Sci USA. 2001;98(20):11720–4. 67. Muizelaar JP, Marmarou A, Young HF, et al. Improving the outcome of
53. Mack WJ, Mocco J, Ducruet AF, et al. A cerebroprotective dose of intrave- severe head injury with the oxygen radical scavenger polyethylene
nous citrate/sorbitol-stabilized dehydroascorbic acid is correlated with glycol-conjugated superoxide dismutase: a phase II trial. J Neurosurg.
increased cerebral ascorbic acid and inhibited lipid peroxidation after 1993;78(3):375–82.
murine reperfused stroke. Neurosurgery. 2006;59(2):383–8 (discussion 68. Wright DW, Yeatts SD, Silbergleit R, et al. Very early administra-
383–388). tion of progesterone for acute traumatic brain injury. N Engl J Med.
54. Ishaq GM, Saidu Y, Bilbis LS, Muhammad SA, Jinjir N, Shehu BB. Effects of 2014;371(26):2457–66.
α-tocopherol and ascorbic acid in the severity and management of trau- 69. Razmkon A, Sadidi A, Sherafat-Kazemzadeh E, et al. Administration of vita-
matic brain injury in albino rats. J Neurosci Rural Pract. 2013;4(3):292–7. min C and vitamin E in severe head injury: a randomized double-blind
55. Yan M, Yang M, Shao W, et al. High-dose ascorbic acid administration controlled trial. Clin Neurosurg. 2011;58(Journal Article):133–7.
improves functional recovery in rats with spinal cord contusion injury. 70. Siqueira IR, Elsner VR, Leite MC, et al. Ascorbate uptake is decreased in the
Spinal cord. 2014;52(11):803–8. hippocampus of ageing rats. Neurochem Int. 2011;58(4):527–32.
56. Sajja RK, Prasad S, Tang S, Kaisar MA, Cucullo L. Blood-brain barrier disrup- 71. Moor E, Shohami E, Kanevsky E, Grigoriadis N, Symeonidou C, Kohen R.
tion in diabetic mice is linked to Nrf2 signaling deficits: Role of ABCB10? Impairment of the ability of the injured aged brain in elevating urate and
Neurosci Lett. 2017;653:152–8. ascorbate. Exp Gerontol. 2006;41(3):303–11.
57. Zhao J, Moore AN, Redell JB, Dash PK. Enhancing expression of Nrf2- 72. Carr A, Wohlrab C, Young P, Bellomo R. Stability of intravenous vitamin C
driven genes protects the blood brain barrier after brain injury. J Neuro- solutions: a technical report. Crit Care Resusc. 2018;20(3):180–1.
sci. 2007;27(38):10240–8. 73. Wilson JX, Jaworski EM, Kulaga A, Dixon SJ. Substrate regula-
58. Kelly PJ, Morrow JD, Ning M, et al. Oxidative stress and matrix metallopro- tion of ascorbate transport activity in astrocytes. Neurochem Res.
teinase-9 in acute ischemic stroke: the Biomarker Evaluation for Antioxi- 1990;15(10):1037–43.
dant Therapies in Stroke (BEAT-Stroke) study. Stroke. 2008;39(1):100–4. 74. Vera JC, Rivas CI, Fischbarg J, Golde DW. Mammalian facilitative hexose
59. Allahtavakoli M, Amin F, Esmaeeli-Nadimi A, Shamsizadeh A, Kazemi- transporters mediate the transport of dehydroascorbic acid. Nature.
Arababadi M, Kennedy D. Ascorbic acid reduces the adverse effects of 1993;364(6432):79–82.
delayed administration of tissue plasminogen activator in a rat stroke
model. Basic Clin Pharmacol Toxicol. 2015;117(5):335–9.

Potrebbero piacerti anche